Fact-checked by Grok 2 weeks ago

Dicer

Dicer is an endoribonuclease enzyme belonging to the RNase III family, essential for processing double-stranded (dsRNA) and precursor microRNAs (pre-miRNAs) into small interfering RNAs (siRNAs) and mature microRNAs (miRNAs), respectively, thereby initiating (RNAi) pathways in eukaryotes. These small RNAs, typically 20–30 nucleotides long with 2-nucleotide 3′ overhangs, are loaded onto proteins within the (RISC) to guide sequence-specific silencing of target mRNAs through cleavage or translational repression. Dicer was first identified in 2001 in as a bidentate responsible for generating ~22-nucleotide siRNAs from dsRNA in RNAi and for maturing small temporal RNAs (stRNAs) like let-7 that regulate developmental timing. Structurally, Dicer adopts an L-shaped architecture conserved across species, featuring key domains including an N-terminal DExD/H-box helicase domain for ATP-dependent unwinding, a central PAZ domain that recognizes the 3′ end of RNA substrates, two RNase III domains (a and b) that form a pseudodimer for precise endonucleolytic cleavage, and a C-terminal double-stranded RNA-binding domain (dsRBD) for substrate anchoring. In mammals, Dicer is a large ~220 kDa protein, while variations exist, such as multiple Dicer paralogs in plants like Arabidopsis thaliana (four isoforms) and insects like Drosophila melanogaster (Dicer-1 for miRNAs and Dicer-2 for siRNAs). The enzyme's helicase domain not only facilitates processive cleavage of long dsRNAs but also contributes to functional specialization, with ancestral roles in antiviral siRNA production evolving into derived miRNA biogenesis in animals. In the RNAi pathway, Dicer cleaves long dsRNAs—often derived from viruses or transposons—into siRNAs that trigger mRNA degradation or transcriptional silencing, providing a key defense mechanism against pathogens. For miRNA biogenesis, Dicer collaborates with in the to process primary miRNA transcripts (pri-miRNAs) into pre-miRNAs, which are then exported to the for Dicer-mediated maturation into miRNAs that fine-tune during development, differentiation, and stress responses. Beyond canonical roles, Dicer exhibits non-endonucleolytic functions, such as nuclear localization for maintenance and involvement in processing other small RNAs like tRNAs and snoRNAs, highlighting its multifaceted housekeeping roles in cellular homeostasis. Dysregulation of Dicer is implicated in various diseases, including cancers where reduced expression correlates with poor prognosis, and developmental disorders due to impaired miRNA-mediated timing control, as evidenced by sterility and morphological defects in C. elegans dcr-1 mutants. Evolutionarily, Dicer is absent in and some parasitic but present in all major eukaryotic lineages, underscoring its ancient origin tied to RNAi-mediated genome defense.

History and Discovery

Initial Identification

The discovery of (RNAi) by and in 1998 provided the foundational context for identifying key enzymes in small RNA pathways, demonstrating that double-stranded RNA (dsRNA) could specifically silence in . This breakthrough, which earned them the 2006 Nobel Prize in Physiology or Medicine, highlighted the need to uncover the molecular machinery processing dsRNA into functional silencing triggers. An early clue to such machinery came in 1994, when a Dicer-like RNase III gene was identified in a screen of laevis ovary cDNA. In 2001, the enzyme was named and first identified in by Bernstein et al. as a member of the RNase III family of endoribonucleases essential for initiating RNAi. Through genetic screening and biochemical assays, they showed that Dicer processes long dsRNA precursors into approximately 22-nucleotide small interfering RNAs (siRNAs), which guide target mRNA degradation. This identification established Dicer as a critical in the RNAi pathway. Concurrently, independent studies linked Dicer orthologs to small RNA processing in other organisms. In C. elegans, Ketting et al. reported in 2001 that the Dicer homolog (dcr-1) is required for both RNAi-mediated silencing and the maturation of small temporal RNAs involved in developmental timing, with assays confirming its cleavage of dsRNA into ~22-nucleotide fragments. In humans, Hutvágner et al. demonstrated the same year that human Dicer functions in the maturation of let-7 microRNA precursors, integrating it into endogenous regulatory networks. These parallel discoveries underscored Dicer's conserved role across species in generating from dsRNA substrates.

Key Milestones and Evolutionary Insights

A pivotal structural milestone in understanding Dicer's RNase III domains came from the 2001 crystal structure of the bacterial RNase III homolog from Aquifex aeolicus, which revealed its homodimeric architecture essential for double-stranded RNA cleavage, contrasting with the monomeric form of eukaryotic Dicer. This bacterial model provided early insights into the catalytic mechanism, showing how the enzyme positions RNA substrates across two subunits for coordinated cuts, influencing subsequent models of Dicer's evolution from prokaryotic ancestors. Building on this, the 2003 knockout studies in mice demonstrated Dicer's essentiality, as Dicer-null embryos exhibited lethality around embryonic day 7.5 due to severe developmental defects and stem cell depletion, underscoring its conserved role in mammalian embryogenesis. Advancements in the leveraged cryo-electron microscopy (cryo-EM) to visualize eukaryotic Dicer complexes. A 2018 cryo-EM structure of human Dicer bound to its cofactor TRBP and a pre-miRNA substrate at approximately 4.5 Å resolution illuminated the spatial arrangement of the and RNase III domains, revealing how TRBP stabilizes the complex for precise RNA loading. Complementary studies in the same decade captured partial assemblies with 2 (Ago2), highlighting cofactor interactions that enhance substrate specificity in the (RISC) loading pathway. Recent progress from 2023 onward has further refined the dicing mechanism; for instance, a 2023 cryo-EM structure of human Dicer-pre-miRNA in a dicing state (at 3.0 Å) unveiled the "gym" mechanism, where the pre-miRNA hairpin dynamically adjusts within a flexible pocket formed by the and PAZ domains to align for selective cleavage. A 2024 study extended this by identifying structural determinants of cleavage selectivity, showing how base-pairing mismatches and loop motifs modulate Dicer's ruler-like measuring from the PAZ domain to produce 22-nucleotide products. Dicer exhibits broad evolutionary conservation across eukaryotes, tracing its origins to bacterial RNase III enzymes, which provided the ancestral double-stranded processing capability, though Dicer itself emerged uniquely in eukaryotes with added domains for biogenesis. It is present in most eukaryotic lineages, including , fungi, and , but has been independently lost in certain parasites such as the apicomplexan , likely due to streamlined genomes or alternative silencing pathways. In , Dicer homologs known as Dicer-like (DCL) proteins have diversified into multiple paralogs (e.g., DCL1 for miRNAs, DCL4 for antiviral siRNAs), reflecting adaptations for specialized roles in development and defense, with early duplications predating the animal-plant split. This conservation highlights Dicer's fundamental role in , with variations underscoring evolutionary pressures from viral threats and developmental needs.

Structure and Domains

Overall Architecture

Dicer is a multidomain III with an approximate molecular weight of 220 kDa in humans, featuring a modular that includes an N-terminal DExD/H-box domain, a domain of function (DUF283), a central platform domain, a PAZ domain, two RNase III domains (RIIIa and RIIIb), and a C-terminal double-stranded RNA-binding domain (dsRBD). This organization positions the catalytic RNase III domains centrally, flanked by regulatory elements that coordinate RNA substrate recognition and processing. Cryo-electron microscopy (cryo-EM) studies from 2016 to 2020 have revealed Dicer's overall shape as an elongated, L-shaped (clamp-like) , approximately 200 Å in length, where the domain forms the base and the PAZ domain extends upward to capture RNA ends. This conformation enables precise measurement of double-stranded (dsRNA) length, with the PAZ domain anchoring the 3' end and the RNase III domains positioning the substrate for symmetric cleavage at a fixed distance of about 21-23 from the end. Allosteric regulation in Dicer involves the domain, which links the PAZ domain to the connector and stabilizes binding, while the domain facilitates initial loading by interacting with loops and undergoing ATP-dependent conformational shifts. Recent 2023-2024 structural insights highlight dynamic conformational changes, such as disengagement during the dicing state, that enhance selectivity by clamping in the catalytic center and preventing off-target processing. Notable species variations include the absence of a second, internal dsRBD in Dicer, unlike in where this additional domain contributes to protein stability and efficient dsRNA handling; this difference influences overall structural integrity and cofactor interactions in mammals.

Functional Domains

Dicer possesses several key functional domains that enable its role in , each contributing specific biochemical activities to substrate recognition, cleavage, and regulation. The of unknown function (DUF283), located between the and PAZ domains, exhibits dsRNA-binding activity that aids in substrate clamping and . The PAZ (Piwi-Argonaute-Zwille) domain, located at the , specifically binds the 3' overhangs of double-stranded RNA (dsRNA) substrates, recognizing two-nucleotide 3' overhangs and measuring the distance to ensure precise cleavage products of approximately nucleotides in length. This anchoring mechanism positions the substrate optimally for endonucleolytic activity, enhancing accuracy in small RNA generation. The catalytic core of Dicer consists of two RNase III domains, RNase IIIa and RNase IIIb, which form an intramolecular heteroduplex to cleave dsRNA substrates. RNase IIIb cleaves the 5' strand, while RNase IIIa cleaves the 3' strand, resulting in duplexes with 5' phosphate and 3' hydroxyl ends characteristic of functional s. These domains coordinate to perform asymmetric cuts, with conserved acidic residues facilitating magnesium-dependent . The N-terminal DExD/H-box domain supports ATP-dependent unwinding and remodeling of RNA substrates, facilitating access to structured precursors and aiding in processive . In mammals, this also contributes to auto-inhibition of the until substrate binding induces conformational changes. A 2024 study confirmed the human Dicer domain's capability for and single-stranded binding. Recent findings reveal that NSUN2-mediated modification on enhances Dicer's activity, promoting of DNA damage-associated R-loops to support repair processes. The C-terminal double-stranded RNA-binding domain (dsRBD) stabilizes interactions with dsRNA substrates, enhancing binding affinity and positioning near the RNase III for efficient processing. Accessory motifs, such as the TRBP-binding region within the helicase domain, recruit cofactors like TAR RNA-binding protein (TRBP), which modulates Dicer's activity and specificity . Regulatory motifs, including phosphorylation sites on serine and threonine residues, fine-tune Dicer's function in response to cellular stresses; for instance, ATM/ATR kinase-dependent activates nuclear Dicer for dsRNA processing, linking it to repair pathways.

Mechanism of Action

Processing of miRNA Precursors

In the canonical miRNA biogenesis pathway, primary miRNAs (pri-miRNAs) are first transcribed in the and processed by the Drosha-DGCR8 complex, which excises the stem-loop structure to generate precursor miRNAs (pre-miRNAs) approximately 60-70 nucleotides long with 2-nucleotide 3' overhangs. These pre-miRNAs are then exported to the via Exportin-5-RanGTP, where Dicer recognizes the characteristic structure and cleaves it precisely to produce a ~22-nucleotide miRNA duplex. This cytoplasmic processing step is essential for maturing endogenous miRNAs, distinguishing it from other Dicer substrates. Dicer functions in a complex with cofactors such as TAR RNA-binding protein (TRBP) or protein activator of PKR (), which enhance pre-miRNA stability, facilitate substrate recognition, and promote loading of the resulting duplex into the (RISC). TRBP, in particular, bridges Dicer and 2 (Ago2), the core RISC component, ensuring efficient transfer of the miRNA duplex while discriminating pre-miRNAs from other cellular RNAs. PACT, often in competition with TRBP, can modulate processing fidelity but generally supports similar roles in stabilizing the complex. The mechanistic precision of Dicer's cleavage relies on its PAZ domain anchoring the pre-miRNA's 3' overhang, establishing a rule-based ~22-nucleotide spacing from this end to the scissile phosphates targeted by the RNase III domains. Recent cryo-electron microscopy structures of Dicer bound to pre-miRNA in a dicing state reveal a "GYM motif" in the pre-miRNA stem, which interacts with Dicer's and RNase IIIa domains to optimize conformation and position the cleavage site accurately before . This structural adaptation ensures high-fidelity excision, minimizing off-target cuts. The output of this processing is a mature miRNA:miRNA* duplex, where the miRNA* (passenger) strand is typically discarded, and the guide strand is retained by Ago2 in RISC to direct through translational repression or mRNA degradation. The PAZ and RNase III domains of Dicer play coordinated roles in this binding and cleavage, as elucidated in prior structural studies.

Generation of siRNAs and Other Substrates

Dicer plays a central role in the biogenesis of small interfering RNAs (siRNAs) by cleaving long double-stranded (dsRNA) precursors, typically derived from viral genomes or transgenic constructs, into 21-23 duplexes. These siRNAs are subsequently loaded into the (RISC), where they guide sequence-specific degradation of complementary mRNA targets, thereby mediating . This process contrasts with miRNA precursor handling but shares the core endonucleolytic activity of Dicer. The cleavage mechanism involves processive cutting, where Dicer initiates at a dsRNA and performs multiple rounds of excision along the scaffold, generating phased siRNAs with uniform spacing. This iterative action ensures efficient production of a population of siRNAs that cover the entire length of the precursor, enhancing the breadth of targeted . Beyond canonical dsRNA, Dicer processes non-canonical substrates such as R-loops—RNA:DNA hybrids formed during transcription—particularly in the context of DNA damage response. Recent evidence from 2025 demonstrates that Dicer cleaves these damage-associated R-loops (DARTs) at double-strand breaks, a process facilitated by the RNA methyltransferase NSUN2, which modifies the RNA component to promote accessibility. However, this cleavage exhibits minimal efficiency compared to canonical substrates like hairpins or dsRNA, relying on NSUN2 for enhancement without altering processing of standard dsRNA. In , Dicer-like 4 (DCL4) specializes in generating 21-nucleotide siRNAs from viral dsRNA, serving as a primary defense mechanism against RNA viruses through phased amplification. Dicer's domain further enables ATP-dependent unwinding of structured RNAs, facilitating access to complex precursors in both and systems.

Roles in Cellular Processes

RNA Interference Pathway

The RNA interference (RNAi) pathway serves as a critical mechanism for post-transcriptional gene regulation in eukaryotes, where Dicer plays a central role by processing precursor RNAs into small interfering RNAs (siRNAs) and (miRNAs) that guide the (RISC) to target messenger RNAs (mRNAs). In the canonical RNAi process, Dicer cleaves double-stranded RNA (dsRNA) substrates or pre-miRNAs into ~21-23 duplexes, which are then loaded into RISC. These small RNAs function as guides within RISC: siRNAs typically enable perfect base-pairing with target mRNAs, leading to endonucleolytic cleavage and degradation, while miRNAs often mediate imperfect pairing, resulting in translational repression or mRNA destabilization. Key components of the RNAi pathway integrate with Dicer's activity to ensure efficient small RNA maturation and function. For miRNA biogenesis, pre-miRNAs are exported from the to the by Exportin-5 in a Ran-GTP-dependent manner, allowing Dicer to access and process them into mature miRNAs. Following Dicer-mediated cleavage, the resulting small RNA duplexes are incorporated into the (Ago) subunit of RISC, where Ago2 provides the endonuclease activity essential for cleaving the passenger strand of the duplex and, in the case of siRNAs, the target mRNA. This loading and activation step ensures that only the guide strand remains active in RISC to direct . The RNAi pathway also incorporates regulatory feedback mechanisms involving Dicer itself. Notably, miR-103 and miR-107, which are Dicer-dependent miRNAs, bind to the 3' (UTR) of the Dicer mRNA, thereby autoregulating Dicer expression to maintain optimal levels of production and prevent pathway overload. Quantitatively, Dicer's activity generates a substantial pool of small RNAs within s, with miRNAs collectively accounting for hundreds of thousands of molecules per mammalian , enabling fine-tuned regulation of approximately 60% of human protein-coding genes through coordinated targeting of their 3' UTRs. This scale underscores Dicer's pivotal contribution to the precision and breadth of RNAi-mediated .

DNA Damage Response and Beyond

Dicer plays a critical role in the DNA damage response by facilitating the cleavage of R-loops associated with DNA damage sites, thereby promoting efficient repair and preventing genomic instability. In a , NSUN2, an m⁵C methyltransferase, was shown to localize to double-strand breaks in a transcription-dependent manner, where it methylates damage-responsive transcripts (DARTs) and interacts with Dicer to enhance the processing of DART-associated R-loops, particularly those forming at stalled replication forks. This mechanism resolves R-loops that could otherwise lead to replication fork collapse and persistent DNA damage; experimental evidence from proximity assays, sequencing, and repair efficiency assays in NSUN2-depleted cells demonstrated delayed double-strand break repair and increased genomic instability when this Dicer-NSUN2 interaction is disrupted. Beyond , Dicer is essential for developmental processes, particularly in and embryogenesis, through its role in miRNA biogenesis. Conditional Dicer in embryonic cells results in viable cells that are proficient in self-renewal but severely defective in into multiple lineages, attributable to the absence of mature miRNAs that regulate developmental . Global Dicer ablation in mice leads to embryonic lethality around E6.5–E7.5, characterized by defects and impaired due to miRNA deficiency, as evidenced by the failure to downregulate genes and accumulate developmental regulators in embryos. In cells, Dicer contributes to transposon silencing by generating endogenous small interfering RNAs (endo-siRNAs) from transcripts, thereby maintaining genomic integrity during . In oocytes, a -driven isoform of Dicer directs the production of endo-siRNAs that target and silence and other , preventing their mobilization; Dicer depletion in these cells leads to derepression of transposon activity and increased genomic mutations. Recent Drosophila studies from 2024 further link Dicer to function, showing that TBPH (the TDP-43 homolog) mutations impair Dicer-mediated miRNA biogenesis, resulting in and motor dysfunction; knockdown of Dicer exacerbates TBPH deficiency-induced locomotor deficits in fly models, highlighting a conserved role in neuronal processing. Extending beyond classical RNAi, Dicer participates in heterochromatin formation and function in fission yeast, where it generates siRNAs that guide modifications for epigenetic silencing. In , Dicer (Dcr1) processes noncoding centromeric transcripts into siRNAs that recruit the RITS complex to pericentromeric repeats, promoting H3K9 methylation and Swi6 binding essential for heterochromatin assembly and proper segregation; dcr1 mutants exhibit defects in centromere silencing and increased chromosome loss. This RNAi-directed pathway ensures centromere cohesion and mitotic fidelity, illustrating Dicer's broader involvement in genome maintenance.

Involvement in Diseases

Cancer

Dicer primarily functions as a tumor suppressor in various cancers, where its promotes tumorigenesis through the loss of mature microRNAs (miRNAs) that normally inhibit oncogenic pathways. In models, deletion of a single Dicer1 in heterozygous mutants leads to a global decrease in miRNA levels, accelerating tumor formation and progression compared to wild-type s. Similarly, in nonepithelial ovarian cancers such as Sertoli-Leydig tumors, recurrent missense mutations in the RNase IIIb domain of DICER1 impair miRNA processing and are identified in up to 60% of cases, contributing to tumor development. These mutations, observed across multiple cancer types, disrupt Dicer's endonuclease activity and underscore its broad suppressive role. In certain oncogenic contexts, however, Dicer exhibits paradoxical behavior, with overexpression associated with enhanced cancer progression and metastasis. In prostate cancer, Dicer levels are upregulated during the transition from prostatic intraepithelial neoplasia to invasive adenocarcinoma and further in metastatic lesions, correlating with increased migratory and invasive potential of tumor cells. This dosage-dependent effect highlights Dicer's pleiotropic role, where moderate overexpression may support metastatic spread by altering miRNA profiles that favor epithelial-to-mesenchymal transition. A 2025 study demonstrated that targeted inhibition of Dicer in tumor-associated macrophages reprograms them from a pro-tumoral M2 phenotype to an anti-tumoral M1 phenotype, reducing immunosuppression and suppressing colorectal cancer liver metastasis in vivo. Specific examples illustrate Dicer's impact in gastrointestinal malignancies, such as , where downregulation of Dicer correlates with advanced disease stages and poor patient prognosis, independent of overall survival metrics. This loss contributes to miRNA dysregulation, including the upregulation of oncogenic miR-21, which promotes proliferation and invasion by targeting tumor suppressors like PTEN. Recent therapeutic advances leverage nanoparticle-based delivery systems to specifically inhibit Dicer in the of solid tumors, skewing polarization toward anti-tumor immunity and enhancing overall immune responses against metastatic lesions.

Neurodegenerative and Eye Disorders

Dicer downregulation has been observed in (ALS) models, where cellular stress and mutations in ALS-causing genes such as TDP-43 lead to impaired miRNA biogenesis through sequestration of Dicer into stress granules, reducing its activity and resulting in decreased mature miRNA levels across various ALS forms. In models, deficiency in TBPH—the fly homolog of TDP-43—reduces Dicer (DCR-1 and DCR-2) mRNA and protein levels, causing motor impairments, shortened lifespan, and cytotoxicity that mimic ALS pathology; for instance, neuronal TBPH knockdown shortens male lifespan to 40-41 days (versus 58-60 days in wild-type) and exacerbates locomotor defects, which are partially rescued by Dicer-1 overexpression. Recent 2025 clinical data from a phase Ib/IIa trial demonstrate that enoxacin, a fluoroquinolone , enhances Dicer activity in ALS patients, leading to increased cell-free miRNA levels as pharmacodynamic biomarkers of boosted miRNA biogenesis; these findings suggest potential to slow disease progression, building on preclinical evidence of improved neuromuscular function, and warrant larger efficacy trials. In eye disorders, germline mutations in DICER1 are associated with the DICER1 tumor predisposition syndrome. Conditional of Dicer1 in mouse lens epithelium disrupts lens morphogenesis and induces corneal defects, primarily through the loss of mature miRNAs including miR-184, which is essential for regulating lens development and epithelial integrity; this results in , cataracts, and impaired fiber cell differentiation. In , reduced Dicer1 expression in contributes to disease risk by impairing miRNA-mediated regulation, leading to accumulation of toxic Alu RNAs that trigger activation and RPE cell death, as observed in —the advanced dry form of AMD.

Viral Pathogenesis and Other Conditions

Dicer plays a central role in the host's antiviral defense by processing double-stranded (dsRNA) intermediates produced during into small interfering RNAs (siRNAs), which are incorporated into the (RISC) to target and degrade viral genomes or transcripts. This mechanism, part of the (RNAi) pathway, is particularly prominent in but has been demonstrated in mammalian cells as well, where Dicer-mediated siRNA production suppresses . In response, many viruses have evolved strategies to evade or inhibit Dicer activity, allowing persistent infection. Viruses such as HIV-1 employ proteins like Tat and Vpr to directly target Dicer for inhibition or proteasomal degradation, thereby reducing siRNA production and facilitating viral spread in infected cells. Similarly, the core protein of (HCV) interacts with Dicer to antagonize its function, impairing the processing of viral dsRNA into antiviral siRNAs and promoting HCV replication in hepatocytes. For , Dicer contributes to protection by cleaving viral dsRNA, and its knockdown in cell models leads to enhanced , underscoring its role in limiting despite the virus's segmented structure. Beyond viral infections, Dicer dysregulation is implicated in autoimmune conditions such as systemic lupus erythematosus (SLE), where single nucleotide polymorphisms in the Dicer gene, particularly the AA at a specific locus, are associated with a reduced risk of disease development compared to other genotypes. In reproductive health, Dicer1 mutations or conditional deletions in the female reproductive tract disrupt processing in , leading to impaired follicular maturation, reduced , and due to defects in oocyte quality and function. Studies in insect models, such as , further illuminate Dicer's antiviral roles and provide insights applicable to human immunity; mutations in the Dicer-2 gene result in heightened susceptibility to RNA viruses like flock house virus, as Dicer-2 is essential for generating antiviral siRNAs and mounting an effective RNAi response.

Therapeutic and Diagnostic Applications

Diagnostic Biomarkers

Dicer expression levels serve as a promising for disease prognosis, particularly in , where altered Dicer mRNA in tumor biopsies has been associated with increased risk of metastasis and poorer patient outcomes. For instance, in , lower Dicer expression correlates with enhanced cell migration and stemness properties that promote metastatic progression. Similarly, in , diminished Dicer transcripts predict recurrence and reduced disease-free survival, whereas in , high Dicer expression is associated with poorer outcomes. Circulating microRNAs (miRNAs) have emerged as indirect proxies for Dicer activity, reflecting disruptions in miRNA biogenesis due to altered Dicer function; elevated or dysregulated cell-free miRNAs in plasma can indicate impaired Dicer-mediated processing in various pathologies. Specific molecular assays have been developed to detect Dicer-related alterations for diagnostic purposes. Quantitative polymerase chain reaction (qPCR) is commonly employed to quantify Dicer1 mRNA expression in biopsies, where reduced levels signal aggressive disease and aid in screening for high-risk cases. For Dicer1 mutations, particularly hotspot variants prevalent in endometrial tumors, targeted sequencing or PCR-based methods confirm changes that impair miRNA and drive oncogenesis. In a 2025 phase Ib/IIa clinical study on (), cell-free miRNAs in plasma and were measured via next-generation sequencing and qPCR to monitor responses to Enoxacin, a Dicer-enhancing agent; treatment led to global increases in miRNA levels, validating these as pharmacodynamic biomarkers for Dicer activity restoration. The primary advantage of Dicer-based biomarkers lies in their potential for non-invasive assessment, enabling blood-based tests that avoid tissue biopsies. In age-related (AMD), Dicer1 deficiency induced by contributes to disease pathogenesis, and genetic variants in Dicer1 have been implicated in retinal degeneration risk, supporting the development of peripheral blood assays for early AMD detection. These approaches offer accessibility and repeatability for monitoring progression in neurodegenerative and ocular disorders. Despite these benefits, limitations persist due to Dicer's tissue-specific expression patterns, which necessitate validation in -relevant contexts to avoid misinterpretation of systemic levels. Variability in Dicer regulation across organs can confound results, requiring integrated multi-omics approaches for accurate diagnostic utility.

Therapeutic Targeting Strategies

Therapeutic strategies targeting Dicer focus on modulating its enzymatic activity to restore or disrupt miRNA biogenesis in contexts. approaches aim to enhance Dicer function in conditions characterized by miRNA deficiency, such as neurodegenerative disorders. Enoxacin, a repurposed fluoroquinolone , acts as a Dicer enhancer by stabilizing the Dicer-TRBP complex, thereby promoting pre-miRNA cleavage and increasing mature miRNA levels. In a 2025 Phase Ib/IIa (REALS1) involving patients with (ALS), oral enoxacin dosing elevated plasma cell-free miRNA levels, serving as a pharmacodynamic of Dicer without significant concerns. This trial demonstrated dose-dependent miRNA upregulation, supporting enoxacin's potential to counteract Dicer impairment linked to TDP-43 pathology in ALS. Inhibition strategies target Dicer to suppress oncogenic miRNAs in cancer, where Dicer overexpression can drive tumor progression. Small molecules and siRNAs have been developed to block Dicer activity selectively. For instance, AC1MMYR2 inhibits Dicer-mediated processing of pre-miR-21, an oncogenic miRNA, leading to reduced miR-21 levels, reversal of epithelial-mesenchymal transition, and suppressed tumor growth in preclinical and models. Similarly, siRNA-mediated Dicer knockdown disrupts miRNA maturation, impairing survival by targeting multiple oncogenic pathways. A 2025 study utilized nanoparticles to deliver Dicer-targeting siRNAs, reprogramming tumor-associated macrophages toward an anti-tumoral phenotype and enhancing immune-mediated clearance in colorectal cancer liver models. Gene therapy offers a direct means to correct Dicer1 genetic defects underlying retinal diseases, where Dicer1 mutations or deficiency contribute to degeneration via impaired miRNA of retinal and . CRISPR-Cas9 gene editing has been explored in preclinical models of inherited retinal dystrophies, and given the of Dicer1 in retinal , it holds potential for correcting Dicer1-related defects, though specific applications remain investigational. (AAV) vectors, particularly AAV2 and AAV8 serotypes, facilitate subretinal or intravitreal delivery of components to target retinal cells efficiently. However, challenges include limited cargo capacity of AAV (limiting full systems), potential immune responses to , and ensuring precise editing without off-target effects in the avascular . Several clinical trials have evaluated Dicer-dependent therapies, highlighting both successes and hurdles. MRX34, a liposomal mimic of the tumor-suppressive miR-34a, relies on Dicer for processing into functional mature miRNA to inhibit ; in a Phase I trial for advanced solid tumors including , it showed partial responses in some patients but was halted in 2016 due to severe immune-related adverse events, such as , resulting in patient deaths. Emerging Dicer agonists for neurodegeneration build on enoxacin's profile, with preclinical data indicating restoration of miRNA biogenesis in and other models; further clinical trials are under evaluation as of 2025 to assess long-term efficacy in slowing neuronal loss.

Dicer-Like Proteins in Eukaryotes

In eukaryotes beyond animals, Dicer-like (DCL) proteins exhibit diverse structures and functions adapted to specific biological contexts, particularly in plants where multiple paralogs enable specialized small RNA pathways. In the model plant Arabidopsis thaliana, four DCL paralogs (DCL1–4) have evolved distinct roles in small RNA biogenesis. DCL1 primarily processes primary microRNAs (miRNAs) into 21-nucleotide mature miRNAs, which are essential for developmental regulation and stress responses. DCL2 and DCL4 generate 21-nucleotide small interfering RNAs (siRNAs), with DCL2 contributing to basal antiviral defense and DCL4 playing a dominant role in antiviral siRNA production against RNA viruses, often in partial redundancy with DCL2. DCL3, in contrast, produces 24-nucleotide siRNAs that direct RNA-dependent DNA methylation and chromatin modifications for heterochromatin formation and transposon silencing. These specializations arise from differences in substrate preferences and subcellular localization, allowing plants to integrate endogenous and exogenous RNA silencing pathways. Recent research in the liverwort , an early-diverging land plant, highlights the conserved yet diversified roles of DCLs in non-vascular . M. polymorpha encodes four DCLs (MpDCL1a, MpDCL1b, MpDCL3, and MpDCL4), with MpDCL1a being indispensable for miRNA biogenesis and ; mutants exhibit severely stunted growth, reduced area (0.167 cm² vs. 1.57 cm² in wild-type at 21 days after ), and defective gemmae formation. MpDCL3 regulates patterning by influencing branching and formation through salicylic acid-auxin antagonism and epigenetic siRNA pathways, while MpDCL4 drives trans-acting siRNA (ta-siRNA) and phased siRNA (phasiRNA) production from loci like MpTAS3, impacting regulation and reproductive organ . These findings underscore DCLs' critical contributions to patterning and phased siRNA-mediated developmental control in bryophytes. Plant DCLs further specialize in timing developmental transitions, such as flowering, by modulating key regulators like the FLOWERING LOCUS T () gene through miRNA and siRNA networks. For instance, DCL1-generated miRNAs, including miR156 and miR172, form a temporal cascade that represses FT expression early in development and promotes it later, ensuring proper photoperiodic flowering responses. This pathway exemplifies how DCL-dependent small RNAs integrate environmental cues with endogenous timing mechanisms to optimize . In fungi and protists, DCL variants often feature reduced domain architectures compared to multicellular eukaryotes, reflecting streamlined RNAi functions. Fungal DCLs, such as the two paralogs (MDL1 and MDL2) in Magnaporthe oryzae, typically retain core RNase III and domains but lack some accessory motifs, enabling roles in quelling (endogenous ) and antiviral defense. In protists like trypanosomes (), the DCL proteins (TbDCL1 and TbDCL2) exhibit atypical domain organization specific to trypanosomatids, with TbDCL1 localized cytoplasmically for siRNA processing of retroposon transcripts and TbDCL2 nuclear for repeat-derived siRNAs; these are essential for endogenous RNAi that fine-tunes stage-specific during parasite transitions.

Comparisons with Homologs

Dicer, a key in eukaryotic pathways, shares its catalytic core with prokaryotic RNase III enzymes but exhibits significant structural and functional divergences that reflect evolutionary adaptations for processing small regulatory RNAs. Bacterial RNase III enzymes are homodimeric proteins, each subunit containing a single RNase III domain and a double-stranded RNA-binding domain (dsRBD), but lacking the PAZ domain and motifs characteristic of eukaryotic Dicers. These prokaryotic homologs primarily cleave (rRNA) precursors and other double-stranded RNAs to generate fragments around 11 base pairs long, rather than the 21-23 small interfering RNAs (siRNAs) or microRNAs (miRNAs) produced by Dicer. This simpler architecture enables efficient maturation of structural RNAs essential for assembly in , without the substrate versatility required for regulatory RNA biogenesis in eukaryotes. In , RNase III homologs are sporadically distributed and structurally simplified compared to their bacterial and eukaryotic counterparts, often featuring only a basic RNase III domain without additional accessory modules. These variants contribute to fundamental processing tasks, such as tRNA maturation in select archaeal species, where they cleave precursor transcripts to generate mature forms, albeit through mechanisms distinct from the complex regulatory roles of Dicer. The rarity and minimalism of archaeal RNase III underscore a divergence from the multi-domain elaboration seen in eukaryotes, highlighting an evolutionary bottleneck in dsRNA processing capabilities within this domain of life. Eukaryotic Dicers display kingdom-specific variations that expand functional specialization beyond prokaryotic ancestors. In animals, Dicer typically exists as a single monomeric protein adapted for both miRNA and siRNA pathways, whereas plants encode multiple paralogous Dicer-like (DCL) proteins, each tailored to distinct RNA types—for instance, Arabidopsis thaliana has four DCLs processing miRNAs or siRNAs of varying lengths. A prominent example is in , where Dicer-1 specializes in miRNA biogenesis using a degenerate, ATP-independent domain for precise precursor cleavage, while Dicer-2 handles siRNA production via a functional, ATP-dependent that unwinds longer dsRNA substrates. These paralogous differences in versus enable compartmentalized RNAi responses, contrasting with the unified, non-paralogous processing in prokaryotes. Key structural divergences in Dicer homologs from certain parasites further illustrate functional constraints. For example, the Dicer from the parasite intestinalis lacks both the domain and C-terminal dsRBD, resulting in a minimal that limits substrate versatility to basic dsRNA cleavage without the unwinding or anchoring capabilities of full-length eukaryotic Dicers. Such absences reduce the enzyme's ability to process diverse precursors, potentially creating vulnerabilities in RNAi machinery that differ from systems and influence evolutionary adaptations for survival.

References

  1. [1]
    Molecular mechanisms of Dicer: endonuclease and enzymatic activity
    May 4, 2017 · It is most well known as the endonuclease that functions in the RNA interference (RNAi) pathway to cleave long double-stranded RNA (dsRNA) ...
  2. [2]
    Dicer structure and function: conserved and evolving features
    Jun 13, 2023 · Major Dicer‐dependent mechanisms are RNA interference (RNAi) and microRNA (miRNA) pathways, which employ distinct types of small RNAs. Small ...Dicer Architecture · Dicer Substrates · Dicer Function At The...
  3. [3]
    Dicer functions in RNA interference and in synthesis of small RNA ...
    Dicer is involved in the maturation of small RNA molecules that regulate developmental timing.<|control11|><|separator|>
  4. [4]
    Crystallographic and modeling studies of RNase III ... - PubMed
    We present the crystal structures of the Aa-RNase III endonuclease domain in its ligand-free form and in complex with Mn(2+).<|separator|>
  5. [5]
    Dicer is essential for mouse development - PubMed
    Loss of Dicer1 lead to lethality early in development, with Dicer1-null embryos ... Mice, Knockout; Molecular Sequence Data; RNA Helicases / genetics; RNA ...
  6. [6]
    Determinants of selectivity in the dicing mechanism - Nature
    Oct 18, 2024 · Our research elucidates the cleavage processes of the RNase III enzyme, DICER, which plays a crucial role in the production of small RNAs.
  7. [7]
    Evolution of Animal and Plant Dicers: Early Parallel Duplications ...
    Dicer is absent from bacteria and archaea but is found throughout ... conserved throughout plant DCL-1 but absent from other DCLs (fig. 7). This ...
  8. [8]
    RNA interference in protozoan parasites and its application
    Parasitic protozoan infections represent a major health burden in ... We also discuss the evolutionary loss of the RNAi pathway in protozoan parasites ...
  9. [9]
  10. [10]
    The human Dicer helicase domain is capable of ATP hydrolysis and ...
    Dec 18, 2024 · Here, we reveal that full-length hDicer, through its helicase domain, hydrolyzes ATP. The ATPase activity of hDicer can only be observed under low-turnover ...<|control11|><|separator|>
  11. [11]
  12. [12]
    The C-terminal dsRNA-binding domain of Drosophila Dicer-2 is ...
    Drosophila Dicer-1 and Dicer-2 and human Dicer have a similar multidomain structure (Fig. 1A): an N-terminal helicase domain; a central, atypical dsRNA-binding ...
  13. [13]
    Dicer structure and function: conserved and evolving features - PMC
    Jun 13, 2023 · The base of the enzyme consists of an N‐terminal helicase domain of the RIG‐I‐like receptor subgroup, which forms a clamp‐like structure near ...
  14. [14]
    A conserved lysine is critical for dsRNA cleavage | PNAS
    Dicer, an RNase III enzyme, initiates RNA interference by processing precursor dsRNAs into mature microRNAs and small-interfering RNAs.Abstract · Sign Up For Pnas Alerts · Results And Discussion
  15. [15]
    Auto-inhibition of Human Dicer by its Internal Helicase Domain - PMC
    These results suggest that the DExD/H-box domain likely disrupts the functionality of the Dicer active site until a structural rearrangement occurs, perhaps ...
  16. [16]
    NSUN2 facilitates DICER cleavage of DNA damage-associated R ...
    Aug 23, 2025 · NSUN2 activity promotes DICER cleavage of DARTs-associated R-loops, which is required for efficient DNA repair. We report a role of the RNA m5C ...
  17. [17]
    Phosphorylated nuclear DICER1 promotes open chromatin state ...
    Jul 26, 2023 · KRAS/ERK pathway phosphorylates DICER1, causing its nuclear translocation, and phosphomimetic Dicer1 contributes to tumorigenesis in mice.
  18. [18]
    Nuclear phosphorylated Dicer processes double-stranded RNA in ...
    Recent work implicated human Dicer in the DNA damage response. Burger et al. show that DNA damage induces phosphorylation of Dicer and promotes DNA repair ...Missing: motifs | Show results with:motifs
  19. [19]
    Overview of MicroRNA Biogenesis, Mechanisms of Actions, and ...
    In this review, we provide an update on canonical and non-canonical miRNA biogenesis pathways and various mechanisms underlying miRNA-mediated gene regulations.
  20. [20]
    DICER: structure, function, and regulation - PMC - PubMed Central
    DICER is widely recognized as a key step in the production of miRNAs and siRNAs, although it also has non-canonical functions such as DNA repair and induction ...
  21. [21]
    TRBP ensures efficient Dicer processing of precursor microRNA in ...
    Dec 9, 2016 · MiRNA biogenesis occurs through a series of enzymatic processing steps, including the cleavage of hairpin RNA (precursor miRNA or pre-miRNA) by ...
  22. [22]
    Differential roles of human Dicer-binding proteins TRBP and PACT ...
    May 9, 2013 · We show here that PACT and TRBP have distinct effects on Dicer-mediated dsRNA processing. In particular, we found that PACT in complex with Dicer inhibits the ...Rna Substrate Preparation · Both Trbp And Pact Form... · Figure 2
  23. [23]
    Structure of the human DICER–pre-miRNA complex in a dicing state
    Feb 22, 2023 · Here we report the cryo-electron microscopy structure of hDICER bound to pre-miRNA in a dicing state and uncover the structural basis of pre-miRNA processing.Missing: precursors mechanism
  24. [24]
    siRNA biogenesis and advances in topically applied dsRNA for ...
    Dec 17, 2020 · Once present in the plant cells, long dsRNA molecules are cleaved by Dicer-like (DCL, RNAse III family) enzymes into 21–24-nt small ...
  25. [25]
    Discovery and Use of Long dsRNA Mediated RNA Interference ... - NIH
    With this pathway, virally produced long dsRNA (dsRNA) is cleaved into short interfering RNA (siRNA) by Dicer and loaded into the RNA-induced silencing complex ...Missing: biogenesis | Show results with:biogenesis
  26. [26]
    Key Mechanistic Principles and Considerations Concerning RNA ...
    Aug 13, 2020 · Production of phased small interfering RNAs (siRNAs) requires a double-stranded (dsRNA) terminus where Dicer will initiate processive cleavage.
  27. [27]
    Dicer-2 - Society for Developmental Biology
    Dec 19, 2024 · Dicer makes staggered cuts in dsRNA to form siRNA ... phased siRNAs, consistent with a defined initiation site for Dicer processing.
  28. [28]
    Exogenous and endogenous dsRNAs perceived by plant Dicer-like ...
    Aug 7, 2023 · In plants, RNase III Dicer-like proteins (DCLs) act as sensors of dsRNAs and process them into short 21- to 24-nucleotide (nt) (s)RNAs.
  29. [29]
    Transient kinetic studies of the antiviral Drosophila Dicer-2 reveal ...
    Mar 31, 2021 · ATP hydrolysis promotes transient unwinding of dsRNA termini followed by slow rewinding, and directional translocation of the enzyme to the ...
  30. [30]
    Origins and Mechanisms of miRNAs and siRNAs - PubMed Central
    During the canonical RNAi pathway, the siRNA guide strand directs RISC to perfectly complementary RNA targets, which are then degraded. RNA degradation is ...
  31. [31]
    Dicers at RISC: The Mechanism of RNAi - ScienceDirect.com
    The pathway of RNA interference starts when Dicer cuts dsRNA into small interfering RNAs (siRNAs) that subsequently target homologous mRNAs for destruction.
  32. [32]
    Exportin-5 mediates the nuclear export of pre-microRNAs and short ...
    We demonstrate that human pre-miRNA nuclear export, and miRNA function, are dependent on Exportin-5. Exportin-5 can bind pre-miRNAs specifically in vitro.
  33. [33]
    Human Argonaute2 Mediates RNA Cleavage Targeted by miRNAs ...
    ... endonuclease activity is exclusively associated with Ago2. Exogenously ... RISC Activity Requires Ago2 but Not Ago1, Ago3, or Ago4. After finding that ...
  34. [34]
    the complexity of the mechanisms regulating Dicer gene expression ...
    This review attempts to fill this gap by summarizing the current knowledge of factors that regulate Dicer gene transcription, primary transcript processing, ...
  35. [35]
    Dynamic and Modularized MicroRNA Regulation and Its Implication ...
    Oct 17, 2017 · In humans, it was estimated that 2,588 miRNAs regulate over 60% of human genes and participate in every aspect of cellular activities in cell ...Results · Prediction Of Mirna... · Methods
  36. [36]
  37. [37]
    Dicer1 functions as a haploinsufficient tumor suppressor - PMC - NIH
    Comparing miRNA profiles in Dicer1 wild-type and heterozygous mutant lung cancer cells, there was a global decrease in steady-state miRNA levels in KPD+/− cells ...
  38. [38]
    Recurrent Somatic DICER1 Mutations in Nonepithelial Ovarian ...
    Dec 21, 2011 · In this study, we report recurrent somatic missense mutations of DICER1 that implicate a defect in miRNA processing, not as a permissive event ...Somatic Mutations In Dicer1 · Dicer1 Mutations In The... · Compound Dicer1 Mutations
  39. [39]
  40. [40]
    Up-Regulation of Dicer, a Component of the MicroRNA Machinery ...
    These results suggest that the increase in Dicer expression parallels the progression of PIN to organ-confined prostate cancer and, furthermore, from locally ...
  41. [41]
    Targeting Dicer reprograms tumor-associated macrophages to ...
    Jun 6, 2025 · Inhibition of Dicer skews macrophages toward an anti-tumoral M1 phenotype, thereby suppressing tumor growth in vivo. These studies highlight ...
  42. [42]
    Role of Dicer as a prognostic predictor for survival in cancer patients
    Down-regulation of Dicer was reported in some studies to be associated with poor prognosis of cancer, like ovarian carcinomas [9], colorectal cancers [10], ...
  43. [43]
    Dysregulated miRNA biogenesis downstream of cellular stress ... - NIH
    Sep 1, 2015 · The consecutive reduction in miRNA levels unifies various forms of ALS, suggesting that the development of novel therapies might focus on DICER ...
  44. [44]
    TDP-43 mutations-induced defects in miRNA biogenesis ... - bioRxiv
    Jul 10, 2024 · Here, we demonstrate that TDP-43 and its Drosophila homolog, TBPH, whose mutations crucially perturb Dicer presentation, result in dysregulated ...
  45. [45]
    Dicer Is Involved in Cytotoxicity and Motor Impairment Induced ... - NIH
    Jun 10, 2025 · We show that TBPH is a key regulator of DCR protein expression, highlighting its conserved role in miRNA dysregulation associated with motor function and ...
  46. [46]
    Cell free miRNAs are pharmacodynamic biomarkers for enhanced ...
    Apr 28, 2025 · Conclusion Enhancing DICER activity by Enoxacin, increases miRNA biogenesis in human patients with ALS. These results encourage testing ...
  47. [47]
    DICER1 Tumor Predisposition - GeneReviews® - NCBI Bookshelf
    Apr 24, 2014 · Retinal abnormalities included pigmentary abnormalities, epiretinal membranes, drusen, and retinitis pigmentosa [Huryn et al 2019]. Dental ...
  48. [48]
    Targeted deletion of Dicer disrupts lens morphogenesis, corneal ...
    Sep 1, 2010 · We studied the roles of Dicer and miRNAs in eye development by conditionally deleting the Dicer gene in the mouse lens and corneal epithelium.Missing: Dicer1 | Show results with:Dicer1
  49. [49]
    Dicer functions transcriptionally and posttranscriptionally in ... - PNAS
    Jan 23, 2019 · Dicer plays a pivotal role in the initiation of RNA silencing by recognizing double-stranded RNAs (dsRNAs) and cleaving them into small RNAs ...
  50. [50]
    Dicer functions as an antiviral system against human adenoviruses ...
    Jun 7, 2016 · Viral dsRNAs derived from RNA viruses are cleaved by Dicer, resulting in virus-derived siRNAs in plants, nematodes and insect cells1,2.
  51. [51]
    HIV-1 Tat interaction with Dicer: requirement for RNA - PMC
    Previously, we observed that over-expressed HIV-1 Tat protein can suppress the processing of small RNAs inside cells. Here, we have investigated the ...
  52. [52]
    HCV core protein interacts with Dicer to antagonize RNA silencing
    The HCV core protein has been shown to inhibit the function of Dicer to process double-stranded RNAs (dsRNAs) into siRNAs. Through deletion analysis, we have ...
  53. [53]
    Dicer is involved in protection against influenza A virus infection
    Thus, RNAi is involved in antiviral response, and Dicer is important for protection against influenza A virus infection.Missing: interaction | Show results with:interaction
  54. [54]
    Single nucleotide polymorphisms in the Dicer gene modify the risk of ...
    The AA genotype of Dicer was correlated with a 0.566-fold decreased risk of SLE compared with that of the AG + GG genotype.
  55. [55]
    Impaired microRNA processing causes corpus luteum insufficiency ...
    Apr 8, 2008 · We used a mouse mutant with a hypomorphic Dicer1 allele (Dicer d/d ) and found that Dicer1 deficiency resulted in female infertility.
  56. [56]
    Essential function in vivo for Dicer-2 in host defense against RNA ...
    Conversely, flies with a loss-of-function mutation in the gene encoding Dicer-2 (Dcr-2) showed enhanced susceptibility to infection by flock house virus, ...
  57. [57]
    Genetic Insight into the Domain Structure and Functions of Dicer ...
    Jan 9, 2021 · The name “Dicer” was first introduced into the scientific world in 2001 by Bernstein, Caudy, Hammond and Hannon [5], and it derived from the ...
  58. [58]
    Ribonuclease III mechanisms of double‐stranded RNA cleavage
    ### Summary of Archaeal RNase III or Dicer Homologs in tRNA or RNA Maturation